Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
2.
PLoS Biol ; 19(12): e3001480, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34914695

RESUMEN

Mutations in leucine-rich repeat kinase 2 (LRRK2) cause autosomal dominant Parkinson disease (PD), while polymorphic LRRK2 variants are associated with sporadic PD. PD-linked mutations increase LRRK2 kinase activity and induce neurotoxicity in vitro and in vivo. The small GTPase Rab8a is a LRRK2 kinase substrate and is involved in receptor-mediated recycling and endocytic trafficking of transferrin, but the effect of PD-linked LRRK2 mutations on the function of Rab8a is poorly understood. Here, we show that gain-of-function mutations in LRRK2 induce sequestration of endogenous Rab8a to lysosomes in overexpression cell models, while pharmacological inhibition of LRRK2 kinase activity reverses this phenotype. Furthermore, we show that LRRK2 mutations drive association of endocytosed transferrin with Rab8a-positive lysosomes. LRRK2 has been nominated as an integral part of cellular responses downstream of proinflammatory signals and is activated in microglia in postmortem PD tissue. Here, we show that iPSC-derived microglia from patients carrying the most common LRRK2 mutation, G2019S, mistraffic transferrin to lysosomes proximal to the nucleus in proinflammatory conditions. Furthermore, G2019S knock-in mice show a significant increase in iron deposition in microglia following intrastriatal LPS injection compared to wild-type mice, accompanied by striatal accumulation of ferritin. Our data support a role of LRRK2 in modulating iron uptake and storage in response to proinflammatory stimuli in microglia.


Asunto(s)
Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteínas de Unión al GTP rab/metabolismo , Anciano , Animales , Transporte Biológico , Cuerpo Estriado , Mutación con Ganancia de Función/genética , Células HEK293 , Humanos , Hierro/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía , Persona de Mediana Edad , Mutación , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Proteínas Serina-Treonina Quinasas , Transferrina/metabolismo , Transferrinas/genética , Transferrinas/metabolismo , Proteínas de Unión al GTP rab/genética
3.
Acta Neuropathol ; 144(1): 81-106, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35596783

RESUMEN

The Excitatory Amino Acid Transporter 2 (EAAT2) accounts for 80% of brain glutamate clearance and is mainly expressed in astrocytic perisynaptic processes. EAAT2 function is finely regulated by endocytic events, recycling to the plasma membrane and degradation. Noteworthy, deficits in EAAT2 have been associated with neuronal excitotoxicity and neurodegeneration. In this study, we show that EAAT2 trafficking is impaired by the leucine-rich repeat kinase 2 (LRRK2) pathogenic variant G2019S, a common cause of late-onset familial Parkinson's disease (PD). In LRRK2 G2019S human brains and experimental animal models, EAAT2 protein levels are significantly decreased, which is associated with elevated gliosis. The decreased expression of the transporter correlates with its reduced functionality in mouse LRRK2 G2019S purified astrocytic terminals and in Xenopus laevis oocytes expressing human LRRK2 G2019S. In LRRK2 G2019S knock-in mouse brain, the correct surface localization of the endogenous transporter is impaired, resulting in its interaction with a plethora of endo-vesicular proteins. Mechanistically, we report that pathogenic LRRK2 kinase activity delays the recycling of the transporter to the plasma membrane via Rabs inactivation, causing its intracellular re-localization and degradation. Taken together, our results demonstrate that pathogenic LRRK2 interferes with the physiology of EAAT2, pointing to extracellular glutamate overload as a possible contributor to neurodegeneration in PD.


Asunto(s)
Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Enfermedad de Parkinson , Sistema de Transporte de Aminoácidos X-AG , Animales , Glutamatos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Ratones , Mutación , Neuronas/patología , Enfermedad de Parkinson/patología
4.
Brain ; 144(5): 1509-1525, 2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-33876242

RESUMEN

Parkinson's disease is characterized by the progressive degeneration of dopaminergic neurons within the substantia nigra pars compacta and the presence of protein aggregates in surviving neurons. The LRRK2 G2019S mutation is one of the major determinants of familial Parkinson's disease cases and leads to late-onset Parkinson's disease with pleomorphic pathology, including α-synuclein accumulation and deposition of protein inclusions. We demonstrated that LRRK2 phosphorylates N-ethylmaleimide sensitive factor (NSF). We observed aggregates containing NSF in basal ganglia specimens from patients with Parkinson's disease carrying the G2019S variant, and in cellular and animal models expressing the LRRK2 G2019S variant. We found that LRRK2 G2019S kinase activity induces the accumulation of NSF in toxic aggregates. Of note, the induction of autophagy cleared NSF aggregation and rescued motor and cognitive impairment observed in aged hG2019S bacterial artificial chromosome (BAC) mice. We suggest that LRRK2 G2019S pathological phosphorylation impacts on NSF biochemical properties, thus causing the formation of cytotoxic protein inclusions.


Asunto(s)
Encéfalo/patología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteínas Sensibles a N-Etilmaleimida/metabolismo , Enfermedad de Parkinson/genética , Agregación Patológica de Proteínas/genética , Animales , Autofagia/fisiología , Humanos , Mutación , Enfermedad de Parkinson/patología , Fosforilación , Agregación Patológica de Proteínas/patología
5.
Hum Mol Genet ; 28(23): 3982-3996, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31626293

RESUMEN

Mutations in LRRK2 cause autosomal dominant and sporadic Parkinson's disease, but the mechanisms involved in LRRK2 toxicity in PD are yet to be fully understood. We found that LRRK2 translocates to the nucleus by binding to seven in absentia homolog (SIAH-1), and in the nucleus it directly interacts with lamin A/C, independent of its kinase activity. LRRK2 knockdown caused nuclear lamina abnormalities and nuclear disruption. LRRK2 disease mutations mostly abolish the interaction with lamin A/C and, similar to LRRK2 knockdown, cause disorganization of lamin A/C and leakage of nuclear proteins. Dopaminergic neurons of LRRK2 G2019S transgenic and LRRK2 -/- mice display decreased circularity of the nuclear lamina and leakage of the nuclear protein 53BP1 to the cytosol. Dopaminergic nigral and cortical neurons of both LRRK2 G2019S and idiopathic PD patients exhibit abnormalities of the nuclear lamina. Our data indicate that LRRK2 plays an essential role in maintaining nuclear envelope integrity. Disruption of this function by disease mutations suggests a novel phosphorylation-independent loss-of-function mechanism that may synergize with other neurotoxic effects caused by LRRK2 mutations.


Asunto(s)
Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Membrana Nuclear/metabolismo , Enfermedad de Parkinson/genética , Animales , Células Cultivadas , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Células HEK293 , Humanos , Lamina Tipo A/metabolismo , Mutación con Pérdida de Función , Ratones , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Fosforilación , Ratas , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
6.
Proc Natl Acad Sci U S A ; 114(50): 13176-13181, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29180403

RESUMEN

α-Synuclein accumulation is a pathological hallmark of Parkinson's disease (PD). Ubiquitinated α-synuclein is targeted to proteasomal or lysosomal degradation. Here, we identify SUMOylation as a major mechanism that counteracts ubiquitination by different E3 ubiquitin ligases and regulates α-synuclein degradation. We report that PIAS2 promotes SUMOylation of α-synuclein, leading to a decrease in α-synuclein ubiquitination by SIAH and Nedd4 ubiquitin ligases, and causing its accumulation and aggregation into inclusions. This was associated with an increase in α-synuclein release from the cells. A SUMO E1 inhibitor, ginkgolic acid, decreases α-synuclein levels by relieving the inhibition exerted on α-synuclein proteasomal degradation. α-Synuclein disease mutants are more SUMOylated compared with the wild-type protein, and this is associated with increased aggregation and inclusion formation. We detected a marked increase in PIAS2 expression along with SUMOylated α-synuclein in PD brains, providing a causal mechanism underlying the up-regulation of α-synuclein SUMOylation in the disease. We also found a significant proportion of Lewy bodies in nigral neurons containing SUMO1 and PIAS2. Our observations suggest that SUMOylation of α-synuclein by PIAS2 promotes α-synuclein aggregation by two mutually reinforcing mechanisms. First, it has a direct proaggregatory effect on α-synuclein. Second, SUMOylation facilitates α-synuclein aggregation by blocking its ubiquitin-dependent degradation pathways and promoting its accumulation. Therefore, inhibitors of α-synuclein SUMOylation provide a strategy to reduce α-synuclein levels and possibly aggregation in PD.


Asunto(s)
Enfermedad de Parkinson/metabolismo , Proteolisis , Sumoilación , alfa-Sinucleína/metabolismo , Animales , Células Cultivadas , Células HEK293 , Humanos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteínas Inhibidoras de STAT Activados/genética , Proteínas Inhibidoras de STAT Activados/metabolismo , Ratas Sprague-Dawley , Salicilatos/farmacología , Sustancia Negra/metabolismo
7.
Mov Disord ; 32(3): 423-432, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27911006

RESUMEN

BACKGROUND: Leucine rich repeat kinase 2 (LRRK2) is a promising target for the treatment of Parkinson's disease; however, little is known about the expression of LRRK2 in human brain and if/how LRRK2 protein levels are altered in Parkinson's disease. OBJECTIVES: We measured the protein levels of LRRK2 as well as its phosphorylation on serines 910, 935, and 973 in the postmortem brain tissue of Parkinson's disease patients and aged controls with and without Lewy bodies. METHODS: LRRK2 and its phosphorylation were measured by immunoblot in brain regions differentially affected in Parkinson's disease (n = 30) as well as subjects with Lewy bodies restricted to the periphery and lower brain stem (n = 25) and matched controls without pathology (n = 25). RESULTS: LRRK2 levels were increased in cases with restricted Lewy bodies, with a 30% increase measured in the substantia nigra. In clinical Parkinson's disease, levels of LRRK2 negatively correlated to disease duration and were comparable with controls. LRRK2 phosphorylation, however, particularly at serine 935, was reduced with clinical Parkinson's disease with a 36% reduction measured in the substantia nigra. CONCLUSIONS: Our data show that LRRK2 phosphorylation is reduced with clinical PD, whereas LRRK2 expression is increased in early potential prodromal stages. These results contribute to a better understanding of the role of LRRK2 in idiopathic Parkinson's disease and may aid efforts aimed at therapeutically targeting the LRRK2 protein. © 2016 International Parkinson and Movement Disorder Society.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Corteza Cerebral/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Cuerpos de Lewy/metabolismo , Enfermedad de Parkinson/metabolismo , Sustancia Negra/metabolismo , Factores de Edad , Anciano , Anciano de 80 o más Años , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Enfermedad de Parkinson/fisiopatología
8.
Adv Exp Med Biol ; 1037: 25-43, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29147901

RESUMEN

In 2003, autosomal recessive loss-of-function mutations were identified in PARK7 gene that caused early-onset Parkinson's disease (PD). The PARK7 gene encodes a conserved protein termed DJ-1. DJ-1 is a ubiquitous protein, and within the brain, it is present in the nucleus and cytoplasm of both neuronal and glial cells. DJ-1 is a multifunctional protein, and numerous studies have ascribed various roles, including antioxidative properties, chaperone function, protease activities, mitochondrial functions and regulation of transcription to the protein. The DJ-1 protein undergoes oxidation and post-translational modifications that are important for its function. Not only is DJ-1 linked to familial PD, but it is also associated with the pathogenic mechanisms of sporadic PD and other neurodegenerative disorders where oxidative stress is implicated. In this chapter we provide an overview on the expression of DJ-1 mRNA and protein in different neurodegenerative disorders and discuss some of its main functions together with DJ-1's potential for neuroprotection.


Asunto(s)
Encéfalo/metabolismo , Expresión Génica , Enfermedades Neurodegenerativas/genética , Proteína Desglicasa DJ-1/genética , Animales , Encéfalo/patología , Humanos , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Proteína Desglicasa DJ-1/metabolismo , Procesamiento Proteico-Postraduccional
9.
Hum Mol Genet ; 23(8): 2055-77, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24282027

RESUMEN

Mutations in LRRK2 cause autosomal dominant Parkinson's disease (PD). LRRK2 encodes a multi-domain protein containing GTPase and kinase domains, and putative protein-protein interaction domains. Familial PD mutations alter the GTPase and kinase activity of LRRK2 in vitro. LRRK2 is suggested to regulate a number of cellular pathways although the underlying mechanisms are poorly understood. To explore such mechanisms, it has proved informative to identify LRRK2-interacting proteins, some of which serve as LRRK2 kinase substrates. Here, we identify common interactions of LRRK2 with members of the dynamin GTPase superfamily. LRRK2 interacts with dynamin 1-3 that mediate membrane scission in clathrin-mediated endocytosis and with dynamin-related proteins that mediate mitochondrial fission (Drp1) and fusion (mitofusins and OPA1). LRRK2 partially co-localizes with endosomal dynamin-1 or with mitofusins and OPA1 at mitochondrial membranes. The subcellular distribution and oligomeric complexes of dynamin GTPases are not altered by modulating LRRK2 in mouse brain, whereas mature OPA1 levels are reduced in G2019S PD brains. LRRK2 enhances mitofusin-1 GTP binding, whereas dynamin-1 and OPA1 serve as modest substrates of LRRK2-mediated phosphorylation in vitro. While dynamin GTPase orthologs are not required for LRRK2-induced toxicity in yeast, LRRK2 functionally interacts with dynamin-1 and mitofusin-1 in cultured neurons. LRRK2 attenuates neurite shortening induced by dynamin-1 by reducing its levels, whereas LRRK2 rescues impaired neurite outgrowth induced by mitofusin-1 potentially by reversing excessive mitochondrial fusion. Our study elucidates novel functional interactions of LRRK2 with dynamin-superfamily GTPases that implicate LRRK2 in the regulation of membrane dynamics important for endocytosis and mitochondrial morphology.


Asunto(s)
Dinamina III/metabolismo , Dinamina II/metabolismo , Dinamina I/metabolismo , Neuroblastoma/metabolismo , Enfermedad de Parkinson/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Western Blotting , Estudios de Casos y Controles , Células Cultivadas , Endocitosis , Femenino , GTP Fosfohidrolasas/metabolismo , Células HEK293 , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Ratones , Ratones Noqueados , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Membranas Mitocondriales/metabolismo , Mutación , Neuritas/metabolismo , Neuroblastoma/patología , Neuronas/citología , Neuronas/metabolismo , Enfermedad de Parkinson/patología , Fosforilación , Dominios y Motivos de Interacción de Proteínas , Ratas , Ratas Sprague-Dawley , Saccharomyces cerevisiae , Técnicas del Sistema de Dos Híbridos
10.
Hum Mol Genet ; 23(17): 4621-38, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24740878

RESUMEN

Mutations in the vacuolar protein sorting 35 homolog (VPS35) gene at the PARK17 locus, encoding a key component of the retromer complex, were recently identified as a new cause of late-onset, autosomal dominant Parkinson's disease (PD). Here we explore the pathogenic consequences of PD-associated mutations in VPS35 using a number of model systems. VPS35 exhibits a broad neuronal distribution throughout the rodent brain, including within the nigrostriatal dopaminergic pathway. In the human brain, VPS35 protein levels and distribution are similar in tissues from control and PD subjects, and VPS35 is not associated with Lewy body pathology. The common D620N missense mutation in VPS35 does not compromise its protein stability or localization to endosomal and lysosomal vesicles, or the vesicular sorting of the retromer cargo, sortilin, SorLA and cation-independent mannose 6-phosphate receptor, in rodent primary neurons or patient-derived human fibroblasts. In yeast we show that PD-linked VPS35 mutations are functional and can normally complement VPS35 null phenotypes suggesting that they do not result in a loss-of-function. In rat primary cortical cultures the overexpression of human VPS35 induces neuronal cell death and increases neuronal vulnerability to PD-relevant cellular stress. In a novel viral-mediated gene transfer rat model, the expression of D620N VPS35 induces the marked degeneration of substantia nigra dopaminergic neurons and axonal pathology, a cardinal pathological hallmark of PD. Collectively, these studies establish that dominant VPS35 mutations lead to neurodegeneration in PD consistent with a gain-of-function mechanism, and support a key role for VPS35 in the development of PD.


Asunto(s)
Neuronas Dopaminérgicas/patología , Mutación/genética , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Enfermedad de Parkinson/genética , Proteínas de Transporte Vesicular/genética , Anciano , Anciano de 80 o más Años , Animales , Encéfalo/metabolismo , Encéfalo/patología , Muerte Celular , Células Cultivadas , Dependovirus/metabolismo , Neuronas Dopaminérgicas/metabolismo , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Persona de Mediana Edad , Enfermedad de Parkinson/patología , Estabilidad Proteica , Transporte de Proteínas , Ratas Sprague-Dawley , Saccharomyces cerevisiae/metabolismo , Estrés Fisiológico , Vesículas Transportadoras/metabolismo
11.
Hum Mol Genet ; 23(18): 4887-905, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-24794857

RESUMEN

Mutations in LRRK2 are one of the primary genetic causes of Parkinson's disease (PD). LRRK2 contains a kinase and a GTPase domain, and familial PD mutations affect both enzymatic activities. However, the signaling mechanisms regulating LRRK2 and the pathogenic effects of familial mutations remain unknown. Identifying the signaling proteins that regulate LRRK2 function and toxicity remains a critical goal for the development of effective therapeutic strategies. In this study, we apply systems biology tools to human PD brain and blood transcriptomes to reverse-engineer a LRRK2-centered gene regulatory network. This network identifies several putative master regulators of LRRK2 function. In particular, the signaling gene RGS2, which encodes for a GTPase-activating protein (GAP), is a key regulatory hub connecting the familial PD-associated genes DJ-1 and PINK1 with LRRK2 in the network. RGS2 expression levels are reduced in the striata of LRRK2 and sporadic PD patients. We identify RGS2 as a novel interacting partner of LRRK2 in vivo. RGS2 regulates both the GTPase and kinase activities of LRRK2. We show in mammalian neurons that RGS2 regulates LRRK2 function in the control of neuronal process length. RGS2 is also protective against neuronal toxicity of the most prevalent mutation in LRRK2, G2019S. We find that RGS2 regulates LRRK2 function and neuronal toxicity through its effects on kinase activity and independently of GTPase activity, which reveals a novel mode of action for GAP proteins. This work identifies RGS2 as a promising target for interfering with neurodegeneration due to LRRK2 mutations in PD patients.


Asunto(s)
Redes Reguladoras de Genes , Neuronas/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas RGS/metabolismo , Animales , Encéfalo/metabolismo , Caenorhabditis elegans/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Neuronas/metabolismo , Proteínas Oncogénicas/metabolismo , Enfermedad de Parkinson/sangre , Proteína Desglicasa DJ-1 , Proteínas Quinasas/metabolismo , Biología de Sistemas/métodos , Transcriptoma
12.
J Cell Sci ; 127(Pt 12): 2647-58, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24762811

RESUMEN

DOK1 regulates platelet-derived growth factor (PDGF)-BB-stimulated glioma cell motility. Mechanisms regulating tumour cell motility are essential for invasion and metastasis. We report here that PDGF-BB-mediated glioma cell invasion and migration are dependent on the adaptor protein downstream of kinase 1 (DOK1). DOK1 is expressed in several glioma cell lines and in tumour biopsies from high-grade gliomas. DOK1 becomes tyrosine phosphorylated upon PDGF-BB stimulation of human glioma cells. Knockdown of DOK1 or expression of a DOK1 mutant (DOK1FF) containing Phe in place of Tyr at residues 362 and 398, resulted in inhibition of both the PDGF-BB-induced tyrosine phosphorylation of p130Cas (also known as BCAR1) and the activation of Rap1. DOK1 colocalises with tyrosine phosphorylated p130Cas at the cell membrane of PDGF-BB-treated cells. Expression of a non-tyrosine-phosphorylatable substrate domain mutant of p130Cas (p130Cas15F) inhibited PDGF-BB-mediated Rap1 activation. Knockdown of DOK1 and Rap1 inhibited PDGF-BB-induced chemotactic cell migration, and knockdown of DOK1 and Rap1 and expression of DOK1FF inhibited PDGF-mediated three-dimensional (3D) spheroid invasion. These data show a crucial role for DOK1 in the regulation of PDGF-BB-mediated tumour cell motility through a p130Cas-Rap1 signalling pathway. [Corrected]


Asunto(s)
Neoplasias Encefálicas/metabolismo , Proteína Sustrato Asociada a CrK/metabolismo , Proteínas de Unión al ADN/fisiología , Glioblastoma/metabolismo , Fosfoproteínas/fisiología , Proteínas Proto-Oncogénicas c-sis/fisiología , Proteínas de Unión al ARN/fisiología , Proteínas de Unión a Telómeros/metabolismo , Becaplermina , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Quimiotaxis , Glioblastoma/patología , Humanos , Invasividad Neoplásica , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Complejo Shelterina , Transducción de Señal , Familia-src Quinasas/metabolismo
13.
Biochemistry ; 54(33): 5198-208, 2015 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-26268594

RESUMEN

Leucine-rich repeat kinase 2 (LRRK2) and tau have been identified as risk factors of Parkinson's disease (PD). As LRRK2 is a kinase and tau is hyperphosphorylated in some LRRK2 mutation carriers of PD patients, the obvious hypothesis is that tau could be a substrate of LRRK2. Previous reports that LRRK2 phosphorylates free tau or tubulin-associated tau provide direct support for this proposition. By comparing LRRK2 with cdk5, we show that wild-type LRRK2 and the G2019S mutant phosphorylate free recombinant full-length tau protein with specific activity 480- and 250-fold lower than cdk5, respectively. More strikingly tau binds to wt LRRK2 or the G2019S mutant 140- or 200-fold more strongly than cdk5. The extremely low activity of LRRK2 but strong binding affinity with tau suggests that LRRK2 may facilitate tau phosphorylation as a scaffold protein rather than as a major tau kinase. This hypothesis is further supported by the observation that (i) cdk5 or tau coimmunoprecipitates with endogenous LRRK2 in SH-SY5Y cells, in mouse brain tissue, and in human PBMCs; (ii) knocking down endogenous LRRK2 by its siRNA in SH-SY5Y cells reduces tau phosphorylation at Ser396 and Ser404; (iii) inhibiting LRRK2 kinase activity by its inhibitors has no effect on tau phosphorylation at these two sites; and (iv) overexpressing wt LRRK2, the G2019S mutant, or the D1994A kinase-dead mutant in SH-SY5Y cells has no effect on tau phosphorylation. Our results suggest that LRRK2 facilitates tau phosphorylation indirectly by recruiting tau or cdk5 rather than by directly phosphorylating tau.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas tau/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Biocatálisis , Encéfalo/metabolismo , Línea Celular Tumoral , Humanos , Cinética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Leucocitos Mononucleares/metabolismo , Ratones , Datos de Secuencia Molecular , Mutación , Enfermedad de Parkinson/genética , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Tubulina (Proteína)/metabolismo
14.
J Biol Chem ; 289(31): 21386-400, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24942733

RESUMEN

Mutations in the gene encoding leucine-rich repeat kinase 2 (LRRK2) are a common genetic cause of Parkinson disease, but the mechanisms whereby LRRK2 is regulated are unknown. Phosphorylation of LRRK2 at Ser(910)/Ser(935) mediates interaction with 14-3-3. Pharmacological inhibition of its kinase activity abolishes Ser(910)/Ser(935) phosphorylation and 14-3-3 binding, and this effect is also mimicked by pathogenic mutations. However, physiological situations where dephosphorylation occurs have not been defined. Here, we show that arsenite or H2O2-induced stresses promote loss of Ser(910)/Ser(935) phosphorylation, which is reversed by phosphatase inhibition. Arsenite-induced dephosphorylation is accompanied by loss of 14-3-3 binding and is observed in wild type, G2019S, and kinase-dead D2017A LRRK2. Arsenite stress stimulates LRRK2 self-association and association with protein phosphatase 1α, decreases kinase activity and GTP binding in vitro, and induces translocation of LRRK2 to centrosomes. Our data indicate that signaling events induced by arsenite and oxidative stress may regulate LRRK2 function.


Asunto(s)
Proteínas 14-3-3/metabolismo , Arsenitos/farmacología , Regulación hacia Abajo/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Estrés Fisiológico , Células HEK293 , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Fosforilación , Unión Proteica
15.
J Neurochem ; 135(6): 1242-56, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26375402

RESUMEN

Leucine-rich repeat kinase 2 (LRRK2) is a causative gene for Parkinson's disease, but the physiological function and the mechanism(s) by which the cellular activity of LRRK2 is regulated are poorly understood. Here, we identified p21-activated kinase 6 (PAK6) as a novel interactor of the GTPase/ROC domain of LRRK2. p21-activated kinases are serine-threonine kinases that serve as targets for the small GTP binding proteins Cdc42 and Rac1 and have been implicated in different morphogenetic processes through remodeling of the actin cytoskeleton such as synapse formation and neuritogenesis. Using an in vivo neuromorphology assay, we show that PAK6 is a positive regulator of neurite outgrowth and that LRRK2 is required for this function. Analyses of post-mortem brain tissue from idiopathic and LRRK2 G2019S carriers reveal an increase in PAK6 activation state, whereas knock-out LRRK2 mice display reduced PAK6 activation and phosphorylation of PAK6 substrates. Taken together, these results support a critical role of LRRK2 GTPase domain in cytoskeletal dynamics in vivo through the novel interactor PAK6, and provide a valuable platform to unravel the mechanism underlying LRRK2-mediated pathophysiology. We propose p21-activated kinase 6 (PAK6) as a novel interactor of leucine-rich repeat kinase 2 (LRRK2), a kinase involved in Parkinson's disease (PD). In health, PAK6 regulates neurite complexity in the brain and LRRK2 is required for its function, (a) whereas PAK6 is aberrantly activated in LRRK2-linked PD brain (b) suggesting that LRRK2 toxicity is mediated by PAK6.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Leucina/genética , Neuritas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Quinasas p21 Activadas/genética , Animales , Encéfalo/metabolismo , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Mamíferos/metabolismo , Ratones , Mutación/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Quinasas p21 Activadas/metabolismo
16.
Hum Mol Genet ; 22(10): 2083-96, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23393160

RESUMEN

Parkin E3 ubiquitin-ligase activity and its role in mitochondria homeostasis are thought to play a role in Parkinson's disease (PD). We now report that AF-6 is a novel parkin interacting protein that modulates parkin ubiquitin-ligase activity and mitochondrial roles. Parkin interacts with the AF-6 PDZ region through its C-terminus. This leads to ubiquitination of cytosolic AF-6 and its degradation by the proteasome. On the other hand, endogenous AF-6 robustly increases parkin translocation and ubiquitin-ligase activity at the mitochondria. Mitochondrial AF-6 is not a parkin substrate, but rather co-localizes with parkin and enhances mitochondria degradation through PINK1/parkin-mediated mitophagy. On the other hand, several parkin and PINK1 juvenile disease-mutants are insensitive to AF-6 effects. AF-6 is present in Lewy bodies and its soluble levels are strikingly decreased in the caudate/putamen and substantia nigra of sporadic PD patients, suggesting that decreased AF-6 levels may contribute to the accumulation of dysfunctional mitochondria in the disease. The identification of AF-6 as a positive modulator of parkin translocation to the mitochondria sheds light on the mechanisms involved in PD and underscores AF-6 as a novel target for future therapeutics.


Asunto(s)
Cinesinas/metabolismo , Mitocondrias/metabolismo , Mutación , Miosinas/metabolismo , Enfermedad de Parkinson/metabolismo , Proteínas Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Núcleo Caudado/metabolismo , Núcleo Caudado/patología , Células HEK293 , Humanos , Cinesinas/genética , Mitocondrias/genética , Mitocondrias/patología , Miosinas/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Proteínas Quinasas/genética , Transporte de Proteínas/genética , Proteolisis , Sustancia Negra/metabolismo , Sustancia Negra/patología , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/genética
17.
Biochim Biophys Acta ; 1833(12): 2900-2910, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23916833

RESUMEN

Leucine Rich Repeat Kinase 2 (LRRK2) is one of the most important genetic contributors to Parkinson's disease. LRRK2 has been implicated in a number of cellular processes, including macroautophagy. To test whether LRRK2 has a role in regulating autophagy, a specific inhibitor of the kinase activity of LRRK2 was applied to human neuroglioma cells and downstream readouts of autophagy examined. The resulting data demonstrate that inhibition of LRRK2 kinase activity stimulates macroautophagy in the absence of any alteration in the translational targets of mTORC1, suggesting that LRRK2 regulates autophagic vesicle formation independent of canonical mTORC1 signaling. This study represents the first pharmacological dissection of the role LRRK2 plays in the autophagy/lysosomal pathway, emphasizing the importance of this pathway as a marker for LRRK2 physiological function. Moreover it highlights the need to dissect autophagy and lysosomal activities in the context of LRRK2 related pathologies with the final aim of understanding their aetiology and identifying specific targets for disease modifying therapies in patients.


Asunto(s)
Autofagia , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/enzimología , Autofagia/efectos de los fármacos , Benzodiazepinonas/farmacología , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Inmunohistoquímica , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Proteínas Asociadas a Microtúbulos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Pirimidinas/farmacología , Ratas , Proteína Sequestosoma-1
18.
Proc Natl Acad Sci U S A ; 108(46): 18666-71, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22065755

RESUMEN

α-Synuclein is central to the pathogenesis of Parkinson disease (PD). Mutations as well as accumulation of α-synuclein promote the death of dopaminergic neurons and the formation of Lewy bodies. α-Synuclein is monoubiquitinated by SIAH, but the regulation and roles of monoubiquitination in α-synuclein biology are poorly understood. We now report that the deubiquitinase USP9X interacts in vivo with and deubiquitinates α-synuclein. USP9X levels are significantly lower in cytosolic fractions of PD substantia nigra and Diffuse Lewy Body disease (DLBD) cortices compared to controls. This was associated to lower deubiquitinase activity toward monoubiquitinated α-synuclein in DLBD cortical extracts. A fraction of USP9X seems to be aggregated in PD and DLBD, as USP9X immunoreactivity is detected in Lewy bodies. Knockdown of USP9X expression promotes accumulation of monoubiquitinated α-synuclein species and enhances the formation of toxic α-synuclein inclusions upon proteolytic inhibition. On the other hand, by manipulating USP9X expression levels in the absence of proteolytic impairment, we demonstrate that monoubiquitination controls the partition of α-synuclein between different protein degradation systems. Deubiquitinated α-synuclein is mostly degraded by autophagy, while monoubiquitinated α-synuclein is preferentially degraded by the proteasome. Moreover, monoubiquitination promotes the degradation of α-synuclein, whereas deubiquitination leads to its accumulation, suggesting that the degradation of deubiquitinated α-synuclein by the autophagy pathway is less efficient than the proteasomal one. Lower levels of cytosolic USP9X and deubiquitinase activity in α-synucleinopathies may contribute to the accumulation and aggregation of monoubiquitinated α-synuclein in Lewy bodies. Our data indicate that monoubiquitination is a key determinant of α-synuclein fate.


Asunto(s)
Regulación de la Expresión Génica , Ubiquitina Tiolesterasa/química , Ubiquitina/química , alfa-Sinucleína/química , Autofagia , Línea Celular Tumoral , Corteza Cerebral/metabolismo , Neuronas Dopaminérgicas/metabolismo , Humanos , Cuerpos de Lewy/metabolismo , Enfermedad por Cuerpos de Lewy/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , ARN Interferente Pequeño/metabolismo
19.
J Parkinsons Dis ; 14(3): 495-506, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38640169

RESUMEN

Background: Parkinson's disease is a progressive neurodegenerative disorder mainly distinguished by sporadic etiology, although a genetic component is also well established. Variants in the LRRK2 gene are associated with both familiar and sporadic disease. We have previously shown that PAK6 and 14-3-3γ protein interact with and regulate the activity of LRRK2. Objective: The aim of this study is to quantify PAK6 and 14-3-3γ in plasma as reliable biomarkers for the diagnosis of both sporadic and LRRK2-linked Parkinson's disease. Methods: After an initial quantification of PAK6 and 14-3-3γ expression by means of Western blot in post-mortem human brains, we verified the presence of the two proteins in plasma by using quantitative ELISA tests. We analyzed samples obtained from 39 healthy subjects, 40 patients with sporadic Parkinson's disease, 50 LRRK2-G2019S non-manifesting carriers and 31 patients with LRRK2-G2019S Parkinson's disease. Results: The amount of PAK6 and 14-3-3γ is significantly different in patients with Parkinson's disease compared to healthy subjects. Moreover, the amount of PAK6 also varies with the presence of the G2019S mutation in the LRRK2 gene. Although the generalized linear models show a low association between the presence of Parkinson's disease and PAK6, the kinase could be added in a broader panel of biomarkers for the diagnosis of Parkinson's disease. Conclusions: Changes of PAK6 and 14-3-3γ amount in plasma represent a shared readout for patients affected by sporadic and LRRK2-linked Parkinson's disease. Overall, they can contribute to the establishment of an extended panel of biomarkers for the diagnosis of Parkinson's disease.


Asunto(s)
Proteínas 14-3-3 , Biomarcadores , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Enfermedad de Parkinson , Quinasas p21 Activadas , Humanos , Enfermedad de Parkinson/sangre , Enfermedad de Parkinson/diagnóstico , Enfermedad de Parkinson/genética , Proteínas 14-3-3/sangre , Masculino , Quinasas p21 Activadas/sangre , Quinasas p21 Activadas/metabolismo , Quinasas p21 Activadas/genética , Femenino , Anciano , Biomarcadores/sangre , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Persona de Mediana Edad , Anciano de 80 o más Años , Estudios Prospectivos , Adulto , Mutación
20.
Neurobiol Dis ; 58: 183-90, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23747310

RESUMEN

Mutations in LRRK2 are the most common genetic cause of Parkinson's disease (PD). The most prevalent LRRK2 mutation is the G2019S coding change, located in the kinase domain of this complex multi-domain protein. The majority of G2019S autopsy cases feature typical Lewy Body pathology with a clinical phenotype almost indistinguishable from idiopathic PD (iPD). Here we have investigated the biochemical characteristics of α-synuclein in G2019S LRRK2 PD post-mortem material, in comparison to pathology-matched iPD. Immunohistochemistry with pS129 α-synuclein antibody showed that the medulla is heavily affected with pathology in G2019S PD whilst the basal ganglia (BG), limbic and frontal cortical regions demonstrated comparable pathology scores between G2019S PD and iPD. Significantly lower levels of the highly aggregated α-synuclein species in urea-SDS fractions were observed in G2019S cases compared to iPD in the BG and limbic cortex. Our data, albeit from a small number of cases, highlight a difference in the biochemical properties of aggregated α-synuclein in G2019S linked PD compared to iPD, despite a similar histopathological presentation. This divergence in solubility is most notable in the basal ganglia, a region that is affected preclinically and is damaged before overt dopaminergic cell death.


Asunto(s)
Encéfalo/metabolismo , Cuerpos de Lewy/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Proteínas Serina-Treonina Quinasas/genética , alfa-Sinucleína/metabolismo , Anciano , Anciano de 80 o más Años , Encéfalo/patología , Femenino , Regulación de la Expresión Génica/genética , Glicina/genética , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Masculino , Enfermedad de Parkinson/clasificación , Cambios Post Mortem , Serina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA